Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
J Clin Invest ; 134(8)2024 Mar 11.
Artigo em Inglês | MEDLINE | ID: mdl-38466355

RESUMO

Craniofacial anomalies, especially midline facial defects, are among the most common birth defects in patients and are associated with increased mortality or require lifelong treatment. During mammalian embryogenesis, specific instructions arising at genetic, signaling, and metabolic levels are important for stem cell behaviors and fate determination, but how these functionally relevant mechanisms are coordinated to regulate craniofacial morphogenesis remain unknown. Here, we report that bone morphogenetic protein (BMP) signaling in cranial neural crest cells (CNCCs) is critical for glycolytic lactate production and subsequent epigenetic histone lactylation, thereby dictating craniofacial morphogenesis. Elevated BMP signaling in CNCCs through constitutively activated ACVR1 (ca-ACVR1) suppressed glycolytic activity and blocked lactate production via a p53-dependent process that resulted in severe midline facial defects. By modulating epigenetic remodeling, BMP signaling-dependent lactate generation drove histone lactylation levels to alter essential genes of Pdgfra, thus regulating CNCC behavior in vitro as well as in vivo. These findings define an axis wherein BMP signaling controls a metabolic/epigenetic cascade to direct craniofacial morphogenesis, thus providing a conceptual framework for understanding the interaction between genetic and metabolic cues operative during embryonic development. These findings indicate potential preventive strategies of congenital craniofacial birth defects via modulating metabolic-driven histone lactylation.


Assuntos
Face , Histonas , Animais , Humanos , Epigênese Genética , Histonas/genética , Histonas/metabolismo , Lactatos/metabolismo , Mamíferos/metabolismo , Morfogênese , Crista Neural
2.
JBMR Plus ; 7(4): e10716, 2023 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-37065628

RESUMO

Craniosynostosis is a congenital anomaly characterized by the premature fusion of cranial sutures. Sutures are a critical connective tissue that regulates bone growth; their aberrant fusion results in abnormal shapes of the head and face. The molecular and cellular mechanisms have been investigated for a long time, but knowledge gaps remain between genetic mutations and mechanisms of pathogenesis for craniosynostosis. We previously demonstrated that the augmentation of bone morphogenetic protein (BMP) signaling through constitutively active BMP type 1A receptor (caBmpr1a) in neural crest cells (NCCs) caused the development of premature fusion of the anterior frontal suture, leading to craniosynostosis in mice. In this study, we demonstrated that ectopic cartilage forms in sutures prior to premature fusion in caBmpr1a mice. The ectopic cartilage is subsequently replaced by bone nodules leading to premature fusion with similar but unique fusion patterns between two neural crest-specific transgenic Cre mouse lines, P0-Cre and Wnt1-Cre mice, which coincides with patterns of premature fusion in each line. Histologic and molecular analyses suggest that endochondral ossification in the affected sutures. Both in vitro and in vivo observations suggest a greater chondrogenic capacity and reduced osteogenic capability of neural crest progenitor cells in mutant lines. These results suggest that the augmentation of BMP signaling alters the cell fate of cranial NCCs toward a chondrogenic lineage to prompt endochondral ossification to prematurely fuse cranial sutures. By comparing P0-Cre;caBmpr1a and Wnt1-Cre;caBmpr1a mice at the stage of neural crest formation, we found more cell death of cranial NCCs in P0-Cre;caBmpr1a than Wnt1-Cre;caBmpr1a mice at the developing facial primordia. These findings may provide a platform for understanding why mutations of broadly expressed genes result in the premature fusion of limited sutures. © 2022 The Authors. JBMR Plus published by Wiley Periodicals LLC on behalf of American Society for Bone and Mineral Research.

3.
Genesis ; 61(1-2): e23509, 2023 03.
Artigo em Inglês | MEDLINE | ID: mdl-36622051

RESUMO

Craniofacial anomalies (CFAs) are a diverse group of disorders affecting the shapes of the face and the head. Malformation of the cranial base in humans leads CFAs, such as midfacial hypoplasia and craniosynostosis. These patients have significant burdens associated with breathing, speaking, and chewing. Invasive surgical intervention is the current primary option to correct these structural deficiencies. Understanding molecular cellular mechanism for craniofacial development would provide novel therapeutic options for CFAs. In this study, we found that enhanced bone morphogenetic protein (BMP) signaling in cranial neural crest cells (NCCs) (P0-Cre;caBmpr1a mice) causes premature fusion of intersphenoid synchondrosis (ISS) resulting in leading to short snouts and hypertelorism. Histological analyses revealed reduction of proliferation and higher cell death in ISS at postnatal day 3. We demonstrated to prevent the premature fusion of ISS in P0-Cre;caBmpr1a mice by injecting a p53 inhibitor Pifithrin-α to the pregnant mother from E15.5 to E18.5, resulting in rescue from short snouts and hypertelorism. We further demonstrated to prevent premature fusion of cranial sutures in P0-Cre;caBmpr1a mice by injecting Pifithrin-α through E8.5 to E18.5. These results suggested that enhanced BMP-p53-induced cell death in cranial NCCs causes premature fusion of ISS and sutures in time-dependent manner.


Assuntos
Anormalidades Craniofaciais , Base do Crânio , Proteínas Morfogenéticas Ósseas/metabolismo , Crista Neural/metabolismo , Crista Neural/patologia , Proliferação de Células , Proteína Supressora de Tumor p53/antagonistas & inibidores , Proteína Supressora de Tumor p53/metabolismo , Masculino , Feminino , Animais , Camundongos , Animais Recém-Nascidos , Transdução de Sinais , Apoptose , Condrócitos/metabolismo , Proteínas Smad/metabolismo , Ligação Proteica , Anormalidades Craniofaciais/metabolismo , Anormalidades Craniofaciais/patologia , Gravidez , Base do Crânio/anormalidades , Base do Crânio/metabolismo , Base do Crânio/patologia , Hipertelorismo/metabolismo , Hipertelorismo/patologia
4.
Genesis ; 59(10): e23450, 2021 10.
Artigo em Inglês | MEDLINE | ID: mdl-34487426

RESUMO

Podoplanin, PDPN, is a mucin-type transmembrane glycoprotein widely expressed in many tissues, including lung, kidney, lymph nodes, and mineralized tissues. Its function is critical for lymphatic formation, differentiation of type I alveolar epithelial lung cells, and for bone response to biomechanical loading. It has previously been shown that Pdpn null mice die at birth due to respiratory failure emphasizing the importance of Pdpn in alveolar lung development. During the course of generation of Pdpn mutant mice, we found that most Pdpn null mice in the 129S6 and C57BL6/J mixed genetic background die at the perinatal stage, similar to previously published studies with Pdpn null mice, while all Pdpn null mice bred with Swiss outbred mice survived. Surviving mutant mice in the 129S6 and C57BL6/J mixed genetic background showed alterations in the osteocyte lacunocanalicular network, especially reduced osteocyte canaliculi in the tibial cortex with increased tibial trabecular bone. However, adult Pdpn null mice in the Swiss outbred background showed no overt differences in their osteocyte lacunocnalicular network, bone density, and no overt differences when challenged with exercise. Together, these data suggest that genetic variations present in the Swiss outbred mice compensate for the loss of function of PDPN in lung, kidney, and bone.


Assuntos
Células Epiteliais Alveolares/metabolismo , Diferenciação Celular/genética , Linfangiogênese/genética , Glicoproteínas de Membrana/genética , Animais , Calcificação Fisiológica/genética , Osso Esponjoso/crescimento & desenvolvimento , Osso Esponjoso/metabolismo , Regulação da Expressão Gênica no Desenvolvimento/genética , Rim/crescimento & desenvolvimento , Pulmão/crescimento & desenvolvimento , Pulmão/metabolismo , Linfonodos/crescimento & desenvolvimento , Camundongos , Osteócitos/metabolismo , Tíbia/crescimento & desenvolvimento , Tíbia/metabolismo
5.
Sci Signal ; 14(665)2021 01 12.
Artigo em Inglês | MEDLINE | ID: mdl-33436499

RESUMO

Cranial neural crest cells (CNCCs) are a population of multipotent stem cells that give rise to craniofacial bone and cartilage during development. Bone morphogenetic protein (BMP) signaling and autophagy have been individually implicated in stem cell homeostasis. Mutations that cause constitutive activation of the BMP type I receptor ACVR1 cause the congenital disorder fibrodysplasia ossificans progressiva (FOP), which is characterized by ectopic cartilage and bone in connective tissues in the trunk and sometimes includes ectopic craniofacial bones. Here, we showed that enhanced BMP signaling through the constitutively activated ACVR1 (ca-ACVR1) in CNCCs in mice induced ectopic cartilage formation in the craniofacial region through an autophagy-dependent mechanism. Enhanced BMP signaling suppressed autophagy by activating mTORC1, thus blocking the autophagic degradation of ß-catenin, which, in turn, caused CNCCs to adopt a chondrogenic identity. Transient blockade of mTORC1, reactivation of autophagy, or suppression of Wnt-ß-catenin signaling reduced ectopic cartilages in ca-Acvr1 mutants. Our results suggest that BMP signaling and autophagy coordinately regulate ß-catenin activity to direct the fate of CNCCs during craniofacial development. These findings may also explain why some patients with FOP develop ectopic bones through endochondral ossification in craniofacial regions.


Assuntos
Receptores de Ativinas Tipo I/metabolismo , Condrogênese , Crista Neural/metabolismo , Transdução de Sinais , Crânio/metabolismo , beta Catenina/metabolismo , Receptores de Ativinas Tipo I/genética , Animais , Proteínas Morfogenéticas Ósseas/metabolismo , Alvo Mecanístico do Complexo 1 de Rapamicina/metabolismo , Camundongos , Miosite Ossificante , Crista Neural/fisiologia , Osteogênese , Proteólise , Crânio/fisiologia
6.
J Musculoskelet Neuronal Interact ; 20(1): 149-159, 2020 03 03.
Artigo em Inglês | MEDLINE | ID: mdl-32131380

RESUMO

OBJECTIVES: Conditional Alk2Q207D-floxed (caALK2fl) mice have previously been used as a model of heterotopic ossification (HO). However, HO formation in this model can be highly variable, and it is unclear which methods reliably induce HO. Hence, these studies report validated methods for reproducibly inducing HO in caALK2fl mice. METHODS: Varying doses of Adex-cre and cardiotoxin (CTX) were injected into the calf muscles of 9, 14, or 28-day-old caALK2fl/- or caALK2fl/fl mice. HO was measured by planar radiography or microCT at 14-28 days post-injury. RESULTS: In 9-day-old caALK2fl/- or caALK2fl/fl mice, single injections of 109 PFU Adex-cre and 0.3 µg of CTX were sufficient to induce extensive HO within 14 days post-injury. In 28-day-old mice, the doses were increased to 5 x 109 PFU Adex-cre and 3.0 µg of CTX to achieve similar consistency, but at a slower rate versus younger mice. Using a crush injury, instead of CTX, also provided consistent induction of HO. Finally, the Type 1 BMPR inhibitor, DMH1, significantly reduced HO formation in 28-day-old caALK2fl/fl mice. CONCLUSIONS: These data illustrate multiple methods for reliable induction of localized HO in the caALK2flmouse that can serve as a starting point for new laboratories utilizing this model.


Assuntos
Receptores de Ativinas Tipo I/genética , Músculo Esquelético/diagnóstico por imagem , Músculo Esquelético/lesões , Ossificação Heterotópica/diagnóstico por imagem , Ossificação Heterotópica/genética , Animais , Cardiotoxinas/toxicidade , Membro Posterior/diagnóstico por imagem , Membro Posterior/lesões , Camundongos , Camundongos Transgênicos , Ossificação Heterotópica/induzido quimicamente , Reprodutibilidade dos Testes
7.
J Vis Exp ; (147)2019 05 10.
Artigo em Inglês | MEDLINE | ID: mdl-31132049

RESUMO

Tissue immunostaining provides highly specific and reliable detection of proteins of interest within a given tissue. Here we describe a complete and simple protocol to detect protein expression during craniofacial morphogenesis/pathogenesis using mouse craniofacial tissues as examples. The protocol consists of preparation and cryosectioning of tissues, indirect immunofluorescence, image acquisition, and quantification. In addition, a method for preparation and cryosectioning of undecalcified hard tissues for immunostaining is described, using craniofacial tissues and long bones as examples. Those methods are key to determine the protein expression and morphological/anatomical changes in various tissues during craniofacial morphogenesis/pathogenesis. They are also applicable to other tissues with appropriate modifications. Knowledge of the histology and high quality of sections are critical to draw scientific conclusions from experimental outcomes. Potential limitations of this methodology include but are not limited to specificity of antibodies and difficulties of quantification, which are also discussed here.


Assuntos
Osso e Ossos/anatomia & histologia , Face/anatomia & histologia , Técnicas Histológicas/métodos , Animais , Embrião de Mamíferos/anatomia & histologia , Cabeça , Camundongos , Fatores de Transcrição SOX9/metabolismo
8.
Genesis ; 56(6-7): e23220, 2018 06.
Artigo em Inglês | MEDLINE | ID: mdl-30134066

RESUMO

Craniosynostosis is defined as congenital premature fusion of one or more cranial sutures. While the genetic basis for about 30% of cases is known, the causative genes for the diverse presentations of the remainder of cases are unknown. The recently discovered cranial suture stem cell population affords an opportunity to identify early signaling pathways that contribute to craniosynostosis. We previously demonstrated that enhanced BMP signaling in neural crest cells (caA3 mutants) leads to premature cranial suture fusion resulting in midline craniosynostosis. Since enhanced mTOR signaling in neural crest cells leads to craniofacial bone lesions, we investigated the extent to which mTOR signaling is involved in the pathogenesis of BMP-mediated craniosynostosis by affecting the suture stem cell population. Our results demonstrate a loss of suture stem cells in the caA3 mutant mice by the newborn stage. We have found increased activation of mTOR signaling in caA3 mutant mice during embryonic stages, but not at the newborn stage. Our study demonstrated that inhibition of mTOR signaling via rapamycin in a time specific manner partially rescued the loss of the suture stem cell population. This study provides insight into how enhanced BMP signaling regulates suture stem cells via mTOR activation.


Assuntos
Craniossinostoses/genética , Sirolimo/farmacologia , Serina-Treonina Quinases TOR/efeitos dos fármacos , Animais , Proteínas Morfogenéticas Ósseas/efeitos dos fármacos , Proteínas Morfogenéticas Ósseas/fisiologia , Suturas Cranianas/embriologia , Craniossinostoses/tratamento farmacológico , Modelos Animais de Doenças , Camundongos , Camundongos Endogâmicos C57BL , Crista Neural/metabolismo , Fenótipo , Transdução de Sinais/efeitos dos fármacos , Sirolimo/metabolismo , Crânio/embriologia
9.
Genesis ; 56(3): e23093, 2018 03.
Artigo em Inglês | MEDLINE | ID: mdl-29411501

RESUMO

BMP signaling plays a critical role in craniofacial development. Augmentation of BMPR1A signaling through neural crest-specific expression of constitutively active Bmpr1a (caBmpr1a) results in craniofacial deformities in mice. To investigate whether deletion of Tak1 may rescue the craniofacial deformities caused by enhanced Smad-dependent signaling through caBMPR1A, we generated embryos to activate transcription of caBmpr1a transgene and ablate Tak1 in neural crest derivatives at the same time. We found that deformities of the double mutant mice showed more severe than those with each single mutation, including median facial cleft and cleft palate. We found higher levels of cell death in the medial nasal and the lateral nasal processes at E10.5 in association with higher levels of p53 in the double mutant embryos. We also found higher levels of pSmad1/5/9 in the lateral nasal processes at E10.5 in the double mutant embryos. Western analyses revealed that double mutant embryos showed similar degrees of upregulation of pSmad1/5/9 with caBmpr1a or Tak1-cKO embryos while the double mutant embryos showed higher levels of phospho-p38 than caBmpr1a or Tak1-cKO embryos at E17.5, but not at E10.5. It suggested that deletion of Tak1 aggravates the craniofacial deformities of the caBmpr1a mutants by increasing p53 and phospho-p38 at different stage of embryogenesis.


Assuntos
Receptores de Proteínas Morfogenéticas Ósseas Tipo I/genética , Morte Celular/genética , Anormalidades Craniofaciais/genética , Estudos de Associação Genética , Genótipo , MAP Quinase Quinase Quinases/genética , Mutação , Animais , Apoptose/genética , Biomarcadores , Receptores de Proteínas Morfogenéticas Ósseas Tipo I/metabolismo , Anormalidades Craniofaciais/diagnóstico , Idade Gestacional , Imuno-Histoquímica , MAP Quinase Quinase Quinases/metabolismo , Camundongos , Camundongos Transgênicos , Fenótipo , Transdução de Sinais , Proteínas Smad/metabolismo , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
10.
Dev Biol ; 429(1): 260-270, 2017 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-28641928

RESUMO

Craniosynostosis is caused by premature fusion of one or more sutures in an infant skull, resulting in abnormal facial features. The molecular and cellular mechanisms by which genetic mutations cause craniosynostosis are incompletely characterized, and many of the causative genes for diverse types of syndromic craniosynostosis have not yet been identified. We previously demonstrated that augmentation of BMP signaling mediated by a constitutively active BMP type IA receptor (ca-BmpR1A) in neural crest cells (ca1A hereafter) causes craniosynostosis and superimposition of heterozygous null mutation of Bmpr1a rescues premature suture fusion (ca1A;1aH hereafter). In this study, we superimposed heterozygous null mutations of the other two BMP type I receptors, Bmpr1b and Acvr1 (ca1A;1bH and ca1A;AcH respectively hereafter) to further dissect involvement of BMP-Smad signaling. Unlike caA1;1aH, ca1A;1bH and ca1A;AcH did not restore the craniosynostosis phenotypes. In our in vivo study, Smad-dependent BMP signaling was decreased to normal levels in mut;1aH mice. However, BMP receptor-regulated Smads (R-Smads; pSmad1/5/9 hereafter) levels were comparable between ca1A, ca1A;1bH and ca1A;AcH mice, and elevated compared to control mice. Bmpr1a, Bmpr1b and Acvr1 null cells were used to examine potential mechanisms underlying the differences in ability of heterozygosity for Bmpr1a vs. Bmpr1b or Acvr1 to rescue the mut phenotype. pSmad1/5/9 level was undetectable in Bmpr1a homozygous null cells while pSmad1/5/9 levels did not decrease in Bmpr1b or Acvr1 homozygous null cells. Taken together, our study indicates that different levels of expression and subsequent activation of Smad signaling differentially contribute each BMP type I receptor to BMP-Smad signaling and craniofacial development. These results also suggest differential involvement of each type 1 receptor in pathogenesis of syndromic craniosynostoses.


Assuntos
Receptores de Proteínas Morfogenéticas Ósseas Tipo I/metabolismo , Proteínas Morfogenéticas Ósseas/metabolismo , Transdução de Sinais , Crânio/embriologia , Crânio/metabolismo , Proteínas Smad/metabolismo , Receptores de Ativinas Tipo I/genética , Receptores de Ativinas Tipo I/metabolismo , Animais , Receptores de Proteínas Morfogenéticas Ósseas Tipo I/genética , Craniossinostoses/genética , Craniossinostoses/patologia , Regulação da Expressão Gênica no Desenvolvimento , Heterozigoto , Camundongos Endogâmicos C57BL , Mutação/genética , Osteoblastos/metabolismo , Fenótipo , Transdução de Sinais/genética , Crânio/anormalidades , Crânio/patologia
11.
Sci Rep ; 6: 24256, 2016 Apr 06.
Artigo em Inglês | MEDLINE | ID: mdl-27048979

RESUMO

We previously found that disruption of two type I BMP receptors, Bmpr1a and Acvr1, respectively, in an osteoblast-specific manner, increased bone mass in mice. BMPR1B, another BMP type I receptor, is also capable of binding to BMP ligands and transduce BMP signaling. However, little is known about the function of BMPR1B in bone. In this study, we investigated the bone phenotype in Bmpr1b null mice and the impacts of loss of Bmpr1b on osteoblasts and osteoclasts. We found that deletion of Bmpr1b resulted in osteopenia in 8-week-old male mice, and the phenotype was transient and gender specific. The decreased bone mass was neither due to the changes in osteoblastic bone formation activity nor osteoclastic bone resorption activity in vivo. In vitro differentiation of Bmpr1b null osteoclasts was increased but resorption activity was decreased. Calvarial pre-osteoblasts from Bmpr1b mutant showed comparable differentiation capability in vitro, while they showed increased BMP-SMAD signaling in culture. Different from calvarial pre-osteoblasts, Bmpr1b mutant bone marrow mesenchymal progenitors showed compromised differentiation in vitro, which may be a reason for the osteopenic phenotype in the mutant mice. In conclusion, our results suggested that BMPR1B plays distinct roles from BMPR1A and ACVR1 in maintaining bone mass and transducing BMP signaling.


Assuntos
Receptores de Ativinas Tipo I/genética , Células da Medula Óssea/citologia , Receptores de Proteínas Morfogenéticas Ósseas Tipo I/genética , Deleção de Genes , Células-Tronco Mesenquimais/citologia , Osteoblastos/citologia , Animais , Peso Corporal , Proteínas Morfogenéticas Ósseas/metabolismo , Osso e Ossos/citologia , Diferenciação Celular , Colágeno/química , Feminino , Homeostase , Homozigoto , Ligantes , Masculino , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Mutação , Osteoclastos/citologia , Osteogênese , Fenótipo , Transdução de Sinais , Microtomografia por Raio-X
12.
Development ; 142(7): 1357-67, 2015 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-25742798

RESUMO

Bone morphogenetic protein (BMP) signaling plays many roles in skull morphogenesis. We have previously reported that enhanced BMP signaling through the BMP type IA receptor (BMPR1A) in cranial neural crest cells causes craniosynostosis during postnatal development. Additionally, we observed that 55% of Bmpr1a mutant mice show neonatal lethality characterized by a distended gastrointestinal tract. Here, we show that severely affected mutants exhibit defective nasal cartilage, failure of fusion between the nasal septum and the secondary palate, and higher levels of phosphorylated SMAD1 and SMAD5 in the nasal tissue. TUNEL demonstrated an increase in apoptosis in both condensing mesenchymal tissues and cartilage of the nasal region in mutants. The levels of p53 (TRP53) tumor suppressor protein were also increased in the same tissue. Injection of pifithrin-α, a chemical inhibitor of p53, into pregnant mice prevented neonatal lethality while concomitantly reducing apoptosis in nasal cartilage primordia, suggesting that enhanced BMP signaling induces p53-mediated apoptosis in the nasal cartilage. The expression of Bax and caspase 3, downstream targets of p53, was increased in the mutants; however, the p53 expression level was unchanged. It has been reported that MDM2 interacts with p53 to promote degradation. We found that the amount of MDM2-p53 complex was decreased in all mutants, and the most severely affected mutants had the largest decrease. Our previous finding that the BMP signaling component SMAD1 prevents MDM2-mediated p53 degradation coupled with our new data indicate that augmented BMP signaling induces p53-mediated apoptosis by prevention of p53 degradation in developing nasal cartilage. Thus, an appropriate level of BMP signaling is required for proper craniofacial morphogenesis.


Assuntos
Apoptose , Proteínas Morfogenéticas Ósseas/metabolismo , Morfogênese , Cartilagens Nasais/embriologia , Crista Neural/metabolismo , Transdução de Sinais , Proteína Supressora de Tumor p53/metabolismo , Animais , Animais Recém-Nascidos , Apoptose/efeitos dos fármacos , Benzotiazóis/farmacologia , Receptores de Proteínas Morfogenéticas Ósseas Tipo I/metabolismo , Núcleo Celular/efeitos dos fármacos , Núcleo Celular/metabolismo , Sobrevivência Celular/efeitos dos fármacos , Condrogênese/efeitos dos fármacos , Perda do Embrião/metabolismo , Perda do Embrião/patologia , Embrião de Mamíferos/efeitos dos fármacos , Embrião de Mamíferos/metabolismo , Embrião de Mamíferos/patologia , Fatores de Crescimento de Fibroblastos/metabolismo , Integrases/metabolismo , Mesoderma/efeitos dos fármacos , Mesoderma/embriologia , Mesoderma/patologia , Camundongos , Morfogênese/efeitos dos fármacos , Mutação/genética , Cartilagens Nasais/anormalidades , Cartilagens Nasais/metabolismo , Cartilagens Nasais/patologia , Mucosa Nasal/metabolismo , Crista Neural/efeitos dos fármacos , Crista Neural/embriologia , Nariz/embriologia , Ligação Proteica/efeitos dos fármacos , Proteólise/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-mdm2/metabolismo , Transdução de Sinais/efeitos dos fármacos , Proteínas Smad/metabolismo , Tolueno/análogos & derivados , Tolueno/farmacologia
13.
ACS Biomater Sci Eng ; 1(11): 1139-1150, 2015 Nov 09.
Artigo em Inglês | MEDLINE | ID: mdl-26985455

RESUMO

Trauma-induced heterotopic ossification (HO) and fibrodysplasia ossificans progressiva (FOP) are acquired and genetic variants of pathological bone formation occurring in soft tissues. Conventional treatment modalities target the inflammatory processes preceding bone formation. We investigated the development of a prophylaxis for heterotopic bone formation by addressing the biological basis for HO - dysregulation in the bone morphogenetic protein (BMP) signaling pathway. We previously reported the synthesis of cationic nanogel nanostructured polymers (NSPs) for efficient delivery of short interfering ribonucleic acids (siRNAs) and targeted gene silencing. Results suggested that nanogel:siRNA weight ratios of 1:1 and 5:1 silenced Runx2 and Osx gene expression in primary mouse osteoblasts with a constitutively active (ca) BMP Receptor 1A (BMPR1A) by the Q233D mutation. Repeated RNAi treatments over 14 days significantly inhibited alkaline phosphatase activity in caBMPR1A osteoblasts. Hydroxyapatite (HA) deposition was diminished over 28 days in culture, though complete suppression of HA deposition was not achieved. Outcome data suggested minimal cytotoxicity of nanogel-based RNAi therapeutics, and the multistage disruption of BMP-induced bone formation processes. This RNAi based approach to impeding osteoblastic differentiation and subsequent bone formation may form the basis of a clinical therapy for heterotopic bone formation.

14.
J Bone Miner Res ; 28(6): 1422-33, 2013 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-23281127

RESUMO

Craniosynostosis describes conditions in which one or more sutures of the infant skull are prematurely fused, resulting in facial deformity and delayed brain development. Approximately 20% of human craniosynostoses are thought to result from gene mutations altering growth factor signaling; however, the molecular mechanisms by which these mutations cause craniosynostosis are incompletely characterized, and the causative genes for diverse types of syndromic craniosynostosis have yet to be identified. Here, we show that enhanced bone morphogenetic protein (BMP) signaling through the BMP type IA receptor (BMPR1A) in cranial neural crest cells, but not in osteoblasts, causes premature suture fusion in mice. In support of a requirement for precisely regulated BMP signaling, this defect was rescued on a Bmpr1a haploinsufficient background, with corresponding normalization of Smad phosphorylation. Moreover, in vivo treatment with LDN-193189, a selective chemical inhibitor of BMP type I receptor kinases, resulted in partial rescue of craniosynostosis. Enhanced signaling of the fibroblast growth factor (FGF) pathway, which has been implicated in craniosynostosis, was observed in both mutant and rescued mice, suggesting that augmentation of FGF signaling is not the sole cause of premature fusion found in this model. The finding that relatively modest augmentation of Smad-dependent BMP signaling leads to premature cranial suture fusion suggests an important contribution of dysregulated BMP signaling to syndromic craniosynostoses and potential strategies for early intervention.


Assuntos
Proteínas Morfogenéticas Ósseas/metabolismo , Craniossinostoses/embriologia , Crista Neural/embriologia , Transdução de Sinais , Proteínas Smad/metabolismo , Animais , Receptores de Proteínas Morfogenéticas Ósseas Tipo I/antagonistas & inibidores , Receptores de Proteínas Morfogenéticas Ósseas Tipo I/genética , Receptores de Proteínas Morfogenéticas Ósseas Tipo I/metabolismo , Proteínas Morfogenéticas Ósseas/genética , Craniossinostoses/genética , Craniossinostoses/metabolismo , Craniossinostoses/patologia , Humanos , Camundongos , Camundongos Transgênicos , Mutação , Crista Neural/metabolismo , Crista Neural/patologia , Pirazóis/farmacologia , Pirimidinas/farmacologia , Proteínas Smad/genética
15.
Artigo em Inglês | MEDLINE | ID: mdl-16121005

RESUMO

Programmed cell death (PCD) is an integral part of plant development. Flower petal usually has the shortest lifetime among all plant organs. There must be a sensitive, tightly controlled PCD in the life cycle of the flower. To understand its mechanism, the ethylene production rate of petals and its correlation with degree of senescence, 1-aminocyclopropane-1-carboxylate (ACC) synthase gene expression, ACC synthase activity and ACC content were determined through the whole flower development period which was arbitrarily divided into five stages depending on appearance of the flower. The results showed that ethylene was not detectable at stages 1 and 2, appeared at stage 3 and increased at stage 5. Transcript of ACC synthase gene did not accumulate at stages 1 and 2, but did so at stages 3-5, and increased gradually at stage 5. ACC synthase activity and ACC content changed in similar way to ethylene production. Ethylene plays a critical role in initiation of rose flower senescence through regulating petal PCD.


Assuntos
Apoptose/fisiologia , Etilenos/metabolismo , Flores/crescimento & desenvolvimento , Liases/genética , Rosa/crescimento & desenvolvimento , Apoptose/genética , Northern Blotting , Flores/genética , Flores/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Regulação Enzimológica da Expressão Gênica , Regulação da Expressão Gênica de Plantas , Liases/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Rosa/genética , Rosa/metabolismo
16.
J Zhejiang Univ Sci ; 4(1): 101-8, 2003.
Artigo em Inglês | MEDLINE | ID: mdl-12656351

RESUMO

The authors tested the contents of ABA (abscisic acid), ZR (zeatin riboside), DHZR (dihydrozeatin riboside) and iPA (isopentenyl adenosine) in leafless and leafy apple trees (Red Fuji/Malus micromalus Makino) during soil drought stress. ABA concentration in drought stressed leafless trees increased significantly compared to the controls. ABA both in roots and xylem rose steadily in the earlier drought stage, reaching a maximum of 1.46 +/- 0.35 nmol g(-1) FW and 117 nmol l(-1) after the 8th day. Similar change patterns of ABA concentration was observed in the leafy trees during soil drought stress; ABA concentrations in roots and xylem sap increased and reached the maximum in the first three days; after 8th day, it decreased slightly, whereas leaf ABA concentration increased steadily in drought stressed plants throughout the duration of the experiment. Between drought stressed and control trees, no significant differences were observed in concentration of ZR and DHZR in both leafless and leafy trees; whereas iPA concentration of the drought stressed leafless and leafy plants decreased markedly in the later stage of drought. These results showed that endogenous ABA originated mainly from the roots in the earlier drought stage, and mainly from the leaves in the later drought stage. Total CTK showed no reduction in the earlier drought stage and decreased in the later drought stage.


Assuntos
Ácido Abscísico/metabolismo , Adenosina/análogos & derivados , Citocininas/metabolismo , Desastres , Isopenteniladenosina/análogos & derivados , Malus/metabolismo , Folhas de Planta/metabolismo , Árvores/metabolismo , Água/metabolismo , Adaptação Fisiológica , Adenosina/metabolismo , Cromatografia Líquida de Alta Pressão , Interpretação Estatística de Dados , Desidratação , Ensaio de Imunoadsorção Enzimática , Isopenteniladenosina/metabolismo , Reguladores de Crescimento de Plantas/metabolismo , Folhas de Planta/efeitos dos fármacos , Raízes de Plantas/efeitos dos fármacos , Raízes de Plantas/metabolismo , Solo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...